Tim Sparwasser1, Luciana Berod



Download 199.24 Kb.
Page5/6
Date02.06.2018
Size199.24 Kb.
#52842
1   2   3   4   5   6

132. Mariathasan S, Weiss DS, Newton K, McBride J, O'Rourke K, Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM (2006) Cryopyrin activates the inflammasome in response to toxins and ATP. Nature 440 (7081):228-232. doi:10.1038/nature04515

133. Greenberg S, Di Virgilio F, Steinberg TH, Silverstein SC (1988) Extracellular nucleotides mediate Ca2+ fluxes in J774 macrophages by two distinct mechanisms. The Journal of biological chemistry 263 (21):10337-10343

134. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E, Perfettini JL, Schlemmer F, Tasdemir E, Uhl M, Genin P, Civas A, Ryffel B, Kanellopoulos J, Tschopp J, Andre F, Lidereau R, McLaughlin NM, Haynes NM, Smyth MJ, Kroemer G, Zitvogel L (2009) Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nature medicine 15 (10):1170-1178. doi:10.1038/nm.2028

135. la Sala A, Ferrari D, Corinti S, Cavani A, Di Virgilio F, Girolomoni G (2001) Extracellular ATP induces a distorted maturation of dendritic cells and inhibits their capacity to initiate Th1 responses. Journal of immunology 166 (3):1611-1617

136. Ivanova EP, Alexeeva YV, Pham DK, Wright JP, Nicolau DV (2006) ATP level variations in heterotrophic bacteria during attachment on hydrophilic and hydrophobic surfaces. International microbiology : the official journal of the Spanish Society for Microbiology 9 (1):37-46

137. Mempin R, Tran H, Chen C, Gong H, Kim Ho K, Lu S (2013) Release of extracellular ATP by bacteria during growth. BMC microbiology 13:301. doi:10.1186/1471-2180-13-301

138. Hironaka I, Iwase T, Sugimoto S, Okuda K, Tajima A, Yanaga K, Mizunoe Y (2013) Glucose triggers ATP secretion from bacteria in a growth-phase-dependent manner. Applied and environmental microbiology 79 (7):2328-2335. doi:10.1128/AEM.03871-12

139. Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, Takeda K (2008) ATP drives lamina propria T(H)17 cell differentiation. Nature 455 (7214):808-812. doi:10.1038/nature07240

140. Neves AR, Castelo-Branco MT, Figliuolo VR, Bernardazzi C, Buongusto F, Yoshimoto A, Nanini HF, Coutinho CM, Carneiro AJ, Coutinho-Silva R, de Souza HS (2014) Overexpression of ATP-activated P2X7 receptors in the intestinal mucosa is implicated in the pathogenesis of Crohn's disease. Inflammatory bowel diseases 20 (3):444-457. doi:10.1097/01.MIB.0000441201.10454.06

141. Castro CN, Freitag J, Berod L, Lochner M, Sparwasser T (2015) Microbe-associated immunomodulatory metabolites: Influence on T cell fate and function. Molecular immunology 68 (2 Pt C):575-584. doi:10.1016/j.molimm.2015.07.025

142. Macfarlane S, Macfarlane GT (2003) Regulation of short-chain fatty acid production. Proc Nutr Soc 62 (1):67-72. doi:10.1079/PNS2002207

143. Roediger WE (1980) Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 21 (9):793-798

144. Cummings JH (1984) Colonic absorption: the importance of short chain fatty acids in man. Scand J Gastroenterol Suppl 93:89-99

145. Cummings JH, Pomare EW, Branch WJ, Naylor CP, Macfarlane GT (1987) Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 28 (10):1221-1227

146. Cummings JH, Macfarlane GT (1991) The control and consequences of bacterial fermentation in the human colon. The Journal of applied bacteriology 70 (6):443-459

147. Ganapathy V, Thangaraju M, Prasad PD, Martin PM, Singh N (2013) Transporters and receptors for short-chain fatty acids as the molecular link between colonic bacteria and the host. Curr Opin Pharmacol 13 (6):869-874. doi:10.1016/j.coph.2013.08.006

148. Frankel WL, Zhang W, Singh A, Klurfeld DM, Don S, Sakata T, Modlin I, Rombeau JL (1994) Mediation of the trophic effects of short-chain fatty acids on the rat jejunum and colon. Gastroenterology 106 (2):375-380

149. Wolever TM, Brighenti F, Royall D, Jenkins AL, Jenkins DJ (1989) Effect of rectal infusion of short chain fatty acids in human subjects. The American journal of gastroenterology 84 (9):1027-1033

150. Canfora EE, Jocken JW, Blaak EE (2015) Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol 11 (10):577-591. doi:10.1038/nrendo.2015.128

151. De Vadder F, Kovatcheva-Datchary P, Goncalves D, Vinera J, Zitoun C, Duchampt A, Backhed F, Mithieux G (2014) Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 156 (1-2):84-96. doi:10.1016/j.cell.2013.12.016

152. Davie JR (2003) Inhibition of histone deacetylase activity by butyrate. The Journal of nutrition 133 (7 Suppl):2485S-2493S

153. Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, Muir AI, Wigglesworth MJ, Kinghorn I, Fraser NJ, Pike NB, Strum JC, Steplewski KM, Murdock PR, Holder JC, Marshall FH, Szekeres PG, Wilson S, Ignar DM, Foord SM, Wise A, Dowell SJ (2003) The Orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. The Journal of biological chemistry 278 (13):11312-11319. doi:10.1074/jbc.M211609200

154. Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD, Mellinger JD, Smith SB, Digby GJ, Lambert NA, Prasad PD, Ganapathy V (2009) GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer research 69 (7):2826-2832. doi:10.1158/0008-5472.CAN-08-4466

155. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M (2003) Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. The Journal of biological chemistry 278 (28):25481-25489. doi:10.1074/jbc.M301403200

156. Nilsson NE, Kotarsky K, Owman C, Olde B (2003) Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short-chain fatty acids. Biochemical and biophysical research communications 303 (4):1047-1052

157. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR (2007) Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences of the United States of America 104 (34):13780-13785. doi:10.1073/pnas.0706625104

158. Treem WR, Ahsan N, Shoup M, Hyams JS (1994) Fecal short-chain fatty acids in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr 18 (2):159-164

159. Mazmanian SK, Round JL, Kasper DL (2008) A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453 (7195):620-625. doi:10.1038/nature07008

160. Round JL, Mazmanian SK (2010) Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proceedings of the National Academy of Sciences of the United States of America 107 (27):12204-12209. doi:10.1073/pnas.0909122107

161. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, Xavier RJ, Teixeira MM, Mackay CR (2009) Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461 (7268):1282-U1119. doi:10.1038/nature08530

162. Sina C, Gavrilova O, Forster M, Till A, Derer S, Hildebrand F, Raabe B, Chalaris A, Scheller J, Rehmann A, Franke A, Ott S, Hasler R, Nikolaus S, Folsch UR, Rose-John S, Jiang HP, Li J, Schreiber S, Rosenstiel P (2009) G protein-coupled receptor 43 is essential for neutrophil recruitment during intestinal inflammation. Journal of immunology 183 (11):7514-7522. doi:10.4049/jimmunol.0900063

163. Fournier BM, Parkos CA (2012) The role of neutrophils during intestinal inflammation. Mucosal immunology 5 (4):354-366. doi:10.1038/mi.2012.24

164. Vinolo MA, Hatanaka E, Lambertucci RH, Newsholme P, Curi R (2009) Effects of short chain fatty acids on effector mechanisms of neutrophils. Cell biochemistry and function 27 (1):48-55. doi:10.1002/cbf.1533

165. Vinolo MA, Rodrigues HG, Hatanaka E, Hebeda CB, Farsky SH, Curi R (2009) Short-chain fatty acids stimulate the migration of neutrophils to inflammatory sites. Clinical science 117 (9):331-338. doi:10.1042/CS20080642

166. Vinolo MA, Ferguson GJ, Kulkarni S, Damoulakis G, Anderson K, Bohlooly YM, Stephens L, Hawkins PT, Curi R (2011) SCFAs induce mouse neutrophil chemotaxis through the GPR43 receptor. PloS one 6 (6):e21205. doi:10.1371/journal.pone.0021205

167. Chassaing B, Aitken JD, Malleshappa M, Vijay-Kumar M (2014) Dextran sulfate sodium (DSS)-induced colitis in mice. Curr Protoc Immunol 104:Unit 15 25. doi:10.1002/0471142735.im1525s104

168. Chang PV, Hao L, Offermanns S, Medzhitov R (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proceedings of the National Academy of Sciences of the United States of America 111 (6):2247-2252. doi:10.1073/pnas.1322269111

169. Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M, Krejsgaard T, Biagi E, Andersen MH, Brigidi P, Odum N, Litman T, Woetmann A (2015) The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci Rep 5:16148. doi:10.1038/srep16148

170. Iraporda C, Errea A, Romanin DE, Cayet D, Pereyra E, Pignataro O, Sirard JC, Garrote GL, Abraham AG, Rumbo M (2015) Lactate and short chain fatty acids produced by microbial fermentation downregulate proinflammatory responses in intestinal epithelial cells and myeloid cells. Immunobiology 220 (10):1161-1169. doi:10.1016/j.imbio.2015.06.004

171. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC, Bassi EJ, Moraes-Vieira PM, Hiyane MI, Rodas AC, Peron JP, Aguiar CF, Reis MA, Ribeiro WR, Valduga CJ, Curi R, Vinolo MA, Ferreira CM, Camara NO (2015) Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. Journal of the American Society of Nephrology : JASN 26 (8):1877-1888. doi:10.1681/ASN.2014030288

172. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504 (7480):451-455. doi:10.1038/nature12726

173. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, Lee JR, Offermanns S, Ganapathy V (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40 (1):128-139. doi:10.1016/j.immuni.2013.12.007

174. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H (2013) Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504 (7480):446-450. doi:10.1038/nature12721

175. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341 (6145):569-573. doi:10.1126/science.1241165

176. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, Li B, Turka LA, Olson EN, Greene MI, Wells AD, Hancock WW (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nature medicine 13 (11):1299-1307. doi:10.1038/nm1652

177. de Zoeten EF, Wang L, Sai H, Dillmann WH, Hancock WW (2010) Inhibition of HDAC9 increases T regulatory cell function and prevents colitis in mice. Gastroenterology 138 (2):583-594. doi:10.1053/j.gastro.2009.10.037

178. Wang LQ, de Zoeten EF, Greene MI, Hancock WW (2009) Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of FOXP3(+) regulatory T cells. Nat Rev Drug Discov 8 (12):969-981. doi:10.1038/nrd3031

179. Beier UH, Wang L, Han R, Akimova T, Liu Y, Hancock WW (2012) Histone deacetylases 6 and 9 and sirtuin-1 control Foxp3+ regulatory T cell function through shared and isoform-specific mechanisms. Science signaling 5 (229):ra45. doi:10.1126/scisignal.2002873

180. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, Kim CH (2015) Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal immunology 8 (1):80-93. doi:10.1038/mi.2014.44

181. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M (2015) Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 18 (7):965-977. doi:10.1038/nn.4030

182. Vinolo MA, Rodrigues HG, Nachbar RT, Curi R (2011) Regulation of inflammation by short chain fatty acids. Nutrients 3 (10):858-876. doi:10.3390/nu3100858

183. Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, Olefsky JM (2010) GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 142 (5):687-698. doi:10.1016/j.cell.2010.07.041

184. Talukdar S, Olefsky JM, Osborn O (2011) Targeting GPR120 and other fatty acid-sensing GPCRs ameliorates insulin resistance and inflammatory diseases. Trends in pharmacological sciences 32 (9):543-550. doi:10.1016/j.tips.2011.04.004

185. Fujita T, Matsuoka T, Honda T, Kabashima K, Hirata T, Narumiya S (2011) A GPR40 agonist GW9508 suppresses CCL5, CCL17, and CXCL10 induction in keratinocytes and attenuates cutaneous immune inflammation. The Journal of investigative dermatology 131 (8):1660-1667. doi:10.1038/jid.2011.123

186. Suzuki M, Takaishi S, Nagasaki M, Onozawa Y, Iino I, Maeda H, Komai T, Oda T (2013) Medium-chain fatty acid-sensing receptor, GPR84, is a proinflammatory receptor. The Journal of biological chemistry 288 (15):10684-10691. doi:10.1074/jbc.M112.420042

187. Blomhoff R, Blomhoff HK (2006) Overview of retinoid metabolism and function. Journal of neurobiology 66 (7):606-630. doi:10.1002/neu.20242

188. Giguere V, Ong ES, Segui P, Evans RM (1987) Identification of a receptor for the morphogen retinoic acid. Nature 330 (6149):624-629. doi:10.1038/330624a0

189. Petkovich M, Brand NJ, Krust A, Chambon P (1987) A human retinoic acid receptor which belongs to the family of nuclear receptors. Nature 330 (6147):444-450. doi:10.1038/330444a0

190. Beijer MR, Kraal G, den Haan JM (2014) Vitamin A and dendritic cell differentiation. Immunology 142 (1):39-45

191. Klebanoff CA, Spencer SP, Torabi-Parizi P, Grainger JR, Roychoudhuri R, Ji Y, Sukumar M, Muranski P, Scott CD, Hall JA, Ferreyra GA, Leonardi AJ, Borman ZA, Wang J, Palmer DC, Wilhelm C, Cai R, Sun J, Napoli JL, Danner RL, Gattinoni L, Belkaid Y, Restifo NP (2013) Retinoic acid controls the homeostasis of pre-cDC-derived splenic and intestinal dendritic cells. The Journal of experimental medicine 210 (10):1961-1976. doi:10.1084/jem.20122508

192. Zeng R, Bscheider M, Lahl K, Lee M, Butcher EC (2016) Generation and transcriptional programming of intestinal dendritic cells: essential role of retinoic acid. Mucosal immunology 9 (1):183-193. doi:10.1038/mi.2015.50

193. Haileselassie Y, Navis M, Vu N, Qazi KR, Rethi B, Sverremark-Ekstrom E (2016) Postbiotic Modulation of Retinoic Acid Imprinted Mucosal-like Dendritic Cells by Probiotic Lactobacillus reuteri 17938 In Vitro. Frontiers in immunology 7:96. doi:10.3389/fimmu.2016.00096

194. Bakdash G, Vogelpoel LT, van Capel TM, Kapsenberg ML, de Jong EC (2015) Retinoic acid primes human dendritic cells to induce gut-homing, IL-10-producing regulatory T cells. Mucosal immunology 8 (2):265-278. doi:10.1038/mi.2014.64

195. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY (2004) Retinoic acid imprints gut-homing specificity on T cells. Immunity 21 (4):527-538. doi:10.1016/j.immuni.2004.08.011

196. Manicassamy S, Ravindran R, Deng J, Oluoch H, Denning TL, Kasturi SP, Rosenthal KM, Evavold BD, Pulendran B (2009) Toll-like receptor 2-dependent induction of vitamin A-metabolizing enzymes in dendritic cells promotes T regulatory responses and inhibits autoimmunity. Nature medicine 15 (4):401-409. doi:10.1038/nm.1925

197. Wang S, Villablanca EJ, De Calisto J, Gomes DC, Nguyen DD, Mizoguchi E, Kagan JC, Reinecker HC, Hacohen N, Nagler C, Xavier RJ, Rossi-Bergmann B, Chen YB, Blomhoff R, Snapper SB, Mora JR (2011) MyD88-dependent TLR1/2 signals educate dendritic cells with gut-specific imprinting properties. Journal of immunology 187 (1):141-150. doi:10.4049/jimmunol.1003740

198. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ (2007) All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. The Journal of experimental medicine 204 (8):1765-1774. doi:10.1084/jem.20070719

199. Kang SG, Lim HW, Andrisani OM, Broxmeyer HE, Kim CH (2007) Vitamin A metabolites induce gut-homing FoxP3+ regulatory T cells. Journal of immunology 179 (6):3724-3733

200. Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y (2007) Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. The Journal of experimental medicine 204 (8):1775-1785. doi:10.1084/jem.20070602

201. Elias KM, Laurence A, Davidson TS, Stephens G, Kanno Y, Shevach EM, O'Shea JJ (2008) Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway. Blood 111 (3):1013-1020. doi:10.1182/blood-2007-06-096438

202. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, Cheroutre H (2007) Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 317 (5835):256-260. doi:10.1126/science.1145697

203. Schambach F, Schupp M, Lazar MA, Reiner SL (2007) Activation of retinoic acid receptor-alpha favours regulatory T cell induction at the expense of IL-17-secreting T helper cell differentiation. European journal of immunology 37 (9):2396-2399. doi:10.1002/eji.200737621

204. De Leenheer AP, Lambert WE, Claeys I (1982) All-trans-retinoic acid: measurement of reference values in human serum by high performance liquid chromatography. Journal of lipid research 23 (9):1362-1367

205. Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, Nishiyama M, Sato S, Tsujimura T, Yamamoto M, Yokota Y, Kiyono H, Miyasaka M, Ishii KJ, Akira S (2008) Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nature immunology 9 (7):769-776. doi:10.1038/ni.1622

206. Cha HR, Chang SY, Chang JH, Kim JO, Yang JY, Kim CH, Kweon MN (2010) Downregulation of Th17 cells in the small intestine by disruption of gut flora in the absence of retinoic acid. Journal of immunology 184 (12):6799-6806. doi:10.4049/jimmunol.0902944

207. Wang C, Kang SG, HogenEsch H, Love PE, Kim CH (2010) Retinoic acid determines the precise tissue tropism of inflammatory Th17 cells in the intestine. Journal of immunology 184 (10):5519-5526. doi:10.4049/jimmunol.0903942

208. Pino-Lagos K, Guo Y, Brown C, Alexander MP, Elgueta R, Bennett KA, De Vries V, Nowak E, Blomhoff R, Sockanathan S, Chandraratna RA, Dmitrovsky E, Noelle RJ (2011) A retinoic acid-dependent checkpoint in the development of CD4+ T cell-mediated immunity. The Journal of experimental medicine 208 (9):1767-1775. doi:10.1084/jem.20102358

209. Hall JA, Cannons JL, Grainger JR, Dos Santos LM, Hand TW, Naik S, Wohlfert EA, Chou DB, Oldenhove G, Robinson M, Grigg ME, Kastenmayer R, Schwartzberg PL, Belkaid Y (2011) Essential role for retinoic acid in the promotion of CD4(+) T cell effector responses via retinoic acid receptor alpha. Immunity 34 (3):435-447. doi:10.1016/j.immuni.2011.03.003

210. Sommer A, Tarwotjo I, Hussaini G, Susanto D (1983) Increased mortality in children with mild vitamin A deficiency. Lancet 2 (8350):585-588

211. Stephens D, Jackson PL, Gutierrez Y (1996) Subclinical vitamin A deficiency: a potentially unrecognized problem in the United States. Pediatric nursing 22 (5):377-389, 456

212. Sommer A, Tarwotjo I, Djunaedi E, West KP, Jr., Loeden AA, Tilden R, Mele L (1986) Impact of vitamin A supplementation on childhood mortality. A randomised controlled community trial. Lancet 1 (8491):1169-1173

213. Imdad A, Yakoob MY, Sudfeld C, Haider BA, Black RE, Bhutta ZA (2011) Impact of vitamin A supplementation on infant and childhood mortality. BMC public health 11 Suppl 3:S20. doi:10.1186/1471-2458-11-S3-S20

214. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, Otipoby KL, Yokota A, Takeuchi H, Ricciardi-Castagnoli P, Rajewsky K, Adams DH, von Andrian UH (2006) Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 314 (5802):1157-1160. doi:10.1126/science.1132742

215. Rudraraju R, Surman SL, Jones BG, Sealy R, Woodland DL, Hurwitz JL (2012) Reduced frequencies and heightened CD103 expression among virus-induced CD8(+) T cells in the respiratory tract airways of vitamin A-deficient mice. Clinical and vaccine immunology : CVI 19 (5):757-765. doi:10.1128/CVI.05576-11

216. Surman SL, Rudraraju R, Sealy R, Jones B, Hurwitz JL (2012) Vitamin A deficiency disrupts vaccine-induced antibody-forming cells and the balance of IgA/IgG isotypes in the upper and lower respiratory tract. Viral immunology 25 (4):341-344. doi:10.1089/vim.2012.0023

217. Surman SL, Jones BG, Rudraraju R, Sealy RE, Hurwitz JL (2014) Intranasal administration of retinyl palmitate with a respiratory virus vaccine corrects impaired mucosal IgA response in the vitamin A-deficient host. Clinical and vaccine immunology : CVI 21 (4):598-601. doi:10.1128/CVI.00757-13

218. Surman SL, Jones BG, Sealy RE, Rudraraju R, Hurwitz JL (2014) Oral retinyl palmitate or retinoic acid corrects mucosal IgA responses toward an intranasal influenza virus vaccine in vitamin A deficient mice. Vaccine 32 (22):2521-2524. doi:10.1016/j.vaccine.2014.03.025

219. DeLuca HF (2004) Overview of general physiologic features and functions of vitamin D. The American journal of clinical nutrition 80 (6 Suppl):1689S-1696S

220. Omdahl JL, Morris HA, May BK (2002) Hydroxylase enzymes of the vitamin D pathway: expression, function, and regulation. Annual review of nutrition 22:139-166. doi:10.1146/annurev.nutr.22.120501.150216

221. Lemire JM, Adams JS, Sakai R, Jordan SC (1984) 1 alpha,25-dihydroxyvitamin D3 suppresses proliferation and immunoglobulin production by normal human peripheral blood mononuclear cells. The Journal of clinical investigation 74 (2):657-661. doi:10.1172/JCI111465

222. Rigby WF, Stacy T, Fanger MW (1984) Inhibition of T lymphocyte mitogenesis by 1,25-dihydroxyvitamin D3 (calcitriol). The Journal of clinical investigation 74 (4):1451-1455. doi:10.1172/JCI111557

223. Bhalla AK, Amento EP, Krane SM (1986) Differential effects of 1,25-dihydroxyvitamin D3 on human lymphocytes and monocyte/macrophages: inhibition of interleukin-2 and augmentation of interleukin-1 production. Cell Immunol 98 (2):311-322

224. Bhalla AK, Amento EP, Clemens TL, Holick MF, Krane SM (1983) Specific high-affinity receptors for 1,25-dihydroxyvitamin D3 in human peripheral blood mononuclear cells: presence in monocytes and induction in T lymphocytes following activation. The Journal of clinical endocrinology and metabolism 57 (6):1308-1310. doi:10.1210/jcem-57-6-1308

225. Provvedini DM, Tsoukas CD, Deftos LJ, Manolagas SC (1983) 1,25-dihydroxyvitamin D3 receptors in human leukocytes. Science 221 (4616):1181-1183


Download 199.24 Kb.

Share with your friends:
1   2   3   4   5   6




The database is protected by copyright ©ininet.org 2024
send message

    Main page